You are on page 1of 3

COMMENTARY

COMMENTARY

Dynamics and clustering of IRE1α during ER stress


T. Kelly Rainbolta and Judith Frydmana,b,1

Cellular integrity is critically dependent on maintain- stress through the unfolded protein response (UPR)
ing protein homeostasis. Proteotoxic stresses chal- (2). The UPR relies on three transmembrane signaling
lenging this balance elicit regulated protective proteins, IRE1, PERK, and ATF6, each of which senses
responses essential for organismal fitness. Approxi- protein misfolding in the ER through a luminal domain
mately one-third of synthesized proteins are secreted and transmits to the cytoplasm and nucleus in order to
from cells and undergo biogenesis in the endoplasmic induce the expression of distinct subsets of ER chap-
reticulum (ER). Upon import into the ER, these proteins erones and other factors that ameliorate ER stress (3).
are engaged by chaperone systems to facilitate either In PNAS, Belyy et al. (4) employ advanced quantitative
folding or triage and removal through pathways of ER imaging strategies to reveal the intricacies of the reg-
quality control (1). The activity of ER quality control ulation of IRE1 during ER stress in the cellular context.
is precisely regulated in response to protein folding The most conserved of the three UPR sensors is
IRE1, which is present in organisms from yeast to
mammals. IRE1 is a transmembrane protein composed
ER Stress of three distinct domains, a sensing domain in the ER
A lumen, connected through a transmembrane segment
to a cytosolic regulatory kinase domain, and finally a
C-terminal RNase domain. Upon ER stress, IRE1 is acti-
vated to ultimately result in the noncanonical splicing of
the mRNA encoding the transcription factor XBP1. After
translation, the resulting XBP1-spliced transcription fac-
IRE1 Activity tor drives the expression of ER chaperones and lipid bio-
synthesis genes as a part of transcriptional program to
resolve protein misfolding in the ER (2). In higher eukary-
otes, IRE1 also down-regulates translation of ER-targeted
B Monomers
Dimers
Small Oligomers
Clusters
Large Oligomers Monomers proteins through direct cleavage of ER-associated
mRNAs in a process known as RIDD (5). Evidence from
ER Lumen yeast and mammalian systems suggests two mecha-
nisms through which IRE1 senses increased levels of
Cytosol
protein misfolding in the ER. The luminal domain in-
cludes a segment that has been shown to bind directly
to the ER Hsp70 chaperone BiP, facilitated by its
cochaperone ERdj4 (6). The IRE1 luminal domain can
Assembly Fusion Dissolution
also bind directly to misfolded proteins in the ER
Fig. 1. (A) In response to acute ER stress, IRE1 rapidly forms many small through an MHC-like peptide binding cleft (7). Mis-
oligomers (small green orbs) throughout the ER membrane. These oligomers
folding sensed through these mechanisms leads to
fuse and assemble into a few large clusters (large green orbs) during the first
hours of ER stress. Upon sustained ER stress, IRE1 clusters are eventually IRE1 activation, which occurs through dimerization at
resolved by dissolution back into the free IRE1 pool in the ER membrane. Cluster interfaces in both its ER luminal and cytosolic domains
assembly closely correlates with the induction of IRE1 activities such as that have been observed in crystal structures (8, 9). Be-
XBP1 splicing while dissolution occurs parallel to the shutoff of the IRE1 arm of yond this, early microscopy studies in yeast and mamma-
the UPR. (B) Depiction of the molecular events occurring during IRE1 cluster
formation. First, IRE1 is activated by assembly into dimers and small oligomers.
lian cells with fluorescently tagged IRE1 demonstrated
Then these oligomers fuse to form large clusters that promote IRE1 activity. the formation of large foci during ER stress, suggesting
Finally, clusters are resolved by dissolution of IRE1 back into free monomers. the formation of higher-order oligomers of IRE1 (Fig. 1A)

a
Department of Biology, Stanford University, Stanford, CA 94305; and bDepartment of Genetics, Stanford University, Stanford, CA 94305
Author contributions: T.K.R. and J.F. wrote the paper.
The authors declare no competing interest.
Published under the PNAS license.
See companion article on page 1533 in issue 3 of volume 117.
Downloaded by guest on January 22, 2021

1
To whom correspondence may be addressed. Email: jfrydman@stanford.edu.
First published February 4, 2020.

3352–3354 | PNAS | February 18, 2020 | vol. 117 | no. 7 www.pnas.org/cgi/doi/10.1073/pnas.1921799117


(10, 11). While the formation of these IRE1 clusters is well docu- exchange may only happen at the boundaries. This is reminis-
mented, their nature has remained ambiguous since they are not cent of the lattices formed by other signaling proteins such as the
amenable to purification for biochemistry or structural studies. bacterial chemotaxis system where stable assembly is thought to
What is the architecture of IRE1 clusters? Are they static or mo- promote receptor cooperativity and enhance system sensitivity
bile? How are they assembled and cleared? (16). Such structural cooperativity could contribute to sustaining
Belyy et al. explore the dynamics of IRE1 oligomers in their cellular the splicing activity of IRE1 during ER stress or perhaps alter
context. To this end, they developed an IRE1α-mNeonGreen re- RIDD substrate selection.
porter leveraging the extreme brightness and improved photostability An interesting feature of IRE1 clusters revealed by the automated
of this next-generation florescent protein. High-resolution confocal imaging approach of Belyy et al. is their extensive motility. Analysis of
microscopy revealed that IRE1 clusters are asymmetric, prompting an
investigation at superresolution using structured illumination micros- In PNAS, Belyy et al. employ advanced quantitative
copy (SIM). SIM imaging of cells stressed with tunicamycin, an inhibitor imaging strategies to reveal the intricacies of
of N-linked glycosylation in the ER, revealed that IRE1 clusters have
complex and intricate architectures including structures forming
the regulation of IRE1 during ER stress in the
loops and coils that localize to branching points in the ER network. cellular context.
Interestingly, colocalization with the general ER membrane marker
Sec61β was incomplete, suggesting that IRE1 clusters may exclude the trajectories of thousands of IRE1 clusters suggests their
other ER components and create an IRE1-rich microdomain. This motion is largely diffusive rather than the result of active
could be accomplished through local perturbation of the ER lipid transport along the cytoskeleton. Perhaps most surprising is the
membrane, for instance by compression of the lipid bilayer in the observation that IRE1 cluster diffusion is largely independent of
ER that has been attributed to the IRE1 oligomer (12). These the size of clusters, suggesting their motion is constrained by other
observations suggest the intriguing possibility that IRE1 creates cellular features.
localized platforms that facilitate its function in sensing ER stress One possibility is that IRE1 favors particular subdomains of the ER.
and inducing the UPR. This is supported by previous observations that IRE1 accumulates
Next, Belyy et al. investigated the dynamics of IRE1 clusters at mitochondria–ER contact sites, a domain of the ER referred to
across the timeline of acute ER stress. To this end, they developed as mitochondrial-associated membranes (MAMs) (17, 18). Enrich-
a clonal U2OS cell line expressing inducible tagged IRE1 and ment of IRE1 clusters to MAMs could facilitate signaling from mi-
leveraged automated imaging and analysis pipelines to move well tochondrial sources for integration during the ER stress response
beyond prior microscopy studies of IRE1 clustering in scope and in order to coordinate adaptation across organelles. Furthermore,
depth. Live imaging of single-cell trajectories of IRE1 clustering IRE1 has also been shown to scaffold filamin A and impact cyto-
revealed significant heterogeneity in response to stress, despite skeleton dynamics through which clustering of IRE1 into discrete
analyzing a clonal cell population. Notably, nearly one-half of ER domains could impact the fate of migrating cells (19). It will be
cells never formed detectable IRE1 clusters. Inherent variability interesting to explore the interactions of clustered IRE1 with other
in stress responses could reflect differences in cellular states, such cellular structures through microscopy or proximity labeling ap-
as cell cycle progression or microenvironment, dictating how cells proaches and further investigate how IRE1 clusters may influence
experience stress; alternatively, they could reflect stochastic organelle cross talk during ER stress.
variability in stress response transcription factor activity (13). The Sustained ER stress eventually leads to a shift in the UPR from a
observed latent diversity in how cells respond to stress suggests prosurvival to procell death state (20). This shift is paralleled by a decline
the intriguing possibility that the UPR is not a monolithic response. in IRE1 activation and XBP1 splicing that correlates with a decrease in
The extent and consequences of such diversity in cellular response clustered IRE1. This down-regulation of IRE1 clusters could either be
to ER stress will be of great interest to further explore. accomplished through selective targeting by degradation pathways or
Following only those cells that formed IRE1 clusters, Belyy dispersal of the clusters and return of IRE1 to the free monomer pool in
et al. investigated the dynamics and life cycle of these structures ER. In order to definitively test the mechanism of IRE1 cluster resolution,
upon prolonged ER stress. This analysis confirms prior observa- the authors employed an optical pulse-chase approach using the pho-
tions that many small IRE1 clusters form initially, and then coalesce toconvertible florescent protein mEos4b fused to IRE1. These photo-
into a few large clusters per cell, which ultimately dissipate during conversion experiments further confirmed that IRE1 clusters are not
sustained ER stress (Fig. 1B). The apparent fusion events occurring liquid-like condensates, since their largely immobile cores remain un-
between IRE1 clusters is reminiscent of the behavior of assemblies mixed upon fusion. Imaging cluster dissolution upon continued stress
undergoing liquid–liquid phase separation (14). In recent years, indicated that IRE1 clusters neither break up into smaller clusters
numerous protein assemblies have been shown to form conden- nor are they degraded. Instead, IRE1 molecules melt back into
sates and coalesce through demixing by liquid–liquid phase sepa- the ER network in a sharp transition indicative of an external
ration to regulate a wide variety of activities such as splicing and regulatory signal (Fig. 1B). The dissolution cue remains to be
signal transduction (15). Common features of phase-separated determined, but likely candidates are PERK-regulated dephos-
structures are their dynamic exchange of monomers with bulk solu- phorylation or the increase in free levels of chaperone ERdj4 on
tion and their fluid droplet-like fusion behaviors. To reveal the na- the luminal face of the ER, which returns IRE1 to an inactive state
ture of IRE1 cluster dynamics, the authors performed fluorescence (6, 21). IRE1 cluster dissolution would terminate the transcriptional
recovery after photobleaching experiments, which surprisingly and likely also RIDD activity of activated IRE1, leaving the cell to
demonstrated that, despite their fusion behavior, IRE1 clusters are return to homeostasis or, in the case of continues stress, to shift the
not phase-separated droplets since most IRE1 monomers in clusters response to additional proapoptotic branches of the UPR.
do not readily exchange with the free pool of IRE1 in the ER. In fact, Therapeutic targeting of IRE1 is gaining great interest to treat
Downloaded by guest on January 22, 2021

only a small percentage of IRE1 in clusters is available for exchange, a wide range of diseases, from cancers such as multiple myeloma
suggesting that perhaps IRE1 forms a lattice-like structure where to metabolic disorders such as diabetes (22). However, as of yet,

Rainbolt and Frydman PNAS | February 18, 2020 | vol. 117 | no. 7 | 3353
small-molecule inhibitors or activators of IRE1 have not success- aid in developing future therapeutic strategies targeting IRE1 to
fully transitioned to the clinic. The revealing characterization of ameliorate a variety of diseases.
IRE1’s life cycle during ER stress presented in Belyy et al. dem-
onstrates the intricate complexity and dynamics regulating
Acknowledgments
IRE1 activity. Combined with the tools the authors have devel- We were supported by grants from the NIH (AG056290 and GM056433) and the
oped, this study provides a springboard to realize a more thorough CHDI Foundation (to J.F.). T.K.R. was supported by a NIH National Research
understanding of the function and regulation of IRE1, which could Service Award (GM120947).

1 I. Braakman, N. J. Bulleid, Protein folding and modification in the mammalian endoplasmic reticulum. Annu. Rev. Biochem. 80, 71–99 (2011).
2 P. Walter, D. Ron, The unfolded protein response: From stress pathway to homeostatic regulation. Science 334, 1081–1086 (2011).
3 A. Korennykh, P. Walter, Structural basis of the unfolded protein response. Annu. Rev. Cell Dev. Biol. 28, 251–277 (2012).
4 V. Belyy, N.-H. Tran, P. Walter, Quantitative microscopy reveals dynamics and fate of clustered IRE1α. Proc. Natl. Acad. Sci. U.S.A. 117, 1533–1542 (2020).
5 M. Maurel, E. Chevet, J. Tavernier, S. Gerlo, Getting RIDD of RNA: IRE1 in cell fate regulation. Trends Biochem. Sci. 39, 245–254 (2014).
6 N. Amin-Wetzel et al., A J-protein co-chaperone recruits BiP to monomerize IRE1 and repress the unfolded protein response. Cell 171, 1625–1637.e13 (2017).
7 G. E. Karagöz et al., An unfolded protein-induced conformational switch activates mammalian IRE1. eLife 6, e30700 (2017).
8 A. V. Korennykh et al., The unfolded protein response signals through high-order assembly of Ire1. Nature 457, 687–693 (2009).
9 J. Zhou et al., The crystal structure of human IRE1 luminal domain reveals a conserved dimerization interface required for activation of the unfolded protein
response. Proc. Natl. Acad. Sci. U.S.A. 103, 14343–14348 (2006).
10 Y. Kimata et al., Two regulatory steps of ER-stress sensor Ire1 involving its cluster formation and interaction with unfolded proteins. J. Cell Biol. 179, 75–86 (2007).
11 H. Li, A. V. Korennykh, S. L. Behrman, P. Walter, Mammalian endoplasmic reticulum stress sensor IRE1 signals by dynamic clustering. Proc. Natl. Acad. Sci. U.S.A.
107, 16113–16118 (2010).
12 K. Halbleib et al., Activation of the unfolded protein response by lipid bilayer stress. Mol. Cell 67, 673–684.e8 (2017).
13 D. Popovic, B. Koch, M. Kueblbeck, J. Ellenberg, L. Pelkmans, Multivariate control of transcript to protein variability in single mammalian cells. Cell Syst. 7, 398–
411.e6 (2018).
14 D. Ricci et al., Clustering of IRE1α depends on sensing ER stress but not on its RNase activity. FASEB J. 33, 9811–9827 (2019).
15 S. Alberti, Phase separation in biology. Curr. Biol. 27, R1097–R1102 (2017).
16 A. H. Erbse, J. J. Falke, The core signaling proteins of bacterial chemotaxis assemble to form an ultrastable complex. Biochemistry 48, 6975–6987 (2009).
17 A. Carreras-Sureda et al., Non-canonical function of IRE1α determines mitochondria-associated endoplasmic reticulum composition to control calcium transfer
and bioenergetics. Nat. Cell Biol. 21, 755–767 (2019).
18 T. Mori, T. Hayashi, E. Hayashi, T. P. Su, Sigma-1 receptor chaperone at the ER-mitochondrion interface mediates the mitochondrion-ER-nucleus signaling for
cellular survival. PLoS One 8, e76941 (2013).
19 H. Urra et al., IRE1α governs cytoskeleton remodelling and cell migration through a direct interaction with filamin A. Nat. Cell Biol. 20, 942–953 (2018).
20 R. Jäger, M. J. Bertrand, A. M. Gorman, P. Vandenabeele, A. Samali, The unfolded protein response at the crossroads of cellular life and death during endoplasmic
reticulum stress. Biol. Cell 104, 259–270 (2012).
21 T. K. Chang et al., Coordination between two branches of the unfolded protein response determines apoptotic cell fate. Mol. Cell 71, 629–636.e5 (2018).
22 C. Hetz, E. Chevet, H. P. Harding, Targeting the unfolded protein response in disease. Nat. Rev. Drug Discov. 12, 703–719 (2013).
Downloaded by guest on January 22, 2021

3354 | www.pnas.org/cgi/doi/10.1073/pnas.1921799117 Rainbolt and Frydman

You might also like